Capturing the dynamics of microbial interactions through individual-specific networks

Longitudinal analysis of multivariate individual-specific microbiome profiles over time or across conditions remains dauntin. Most statistical tools and methods that are available to study microbiomes are based on cross-sectional data. Over the past few years, several attempts have been made to mode...

Full description

Bibliographic Details
Main Authors: Behnam Yousefi, Federico Melograna, Gianluca Galazzo, Niels van Best, Monique Mommers, John Penders, Benno Schwikowski, Kristel Van Steen
Format: Article
Language:English
Published: Frontiers Media S.A. 2023-05-01
Series:Frontiers in Microbiology
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fmicb.2023.1170391/full
_version_ 1797826832426336256
author Behnam Yousefi
Behnam Yousefi
Behnam Yousefi
Federico Melograna
Gianluca Galazzo
Niels van Best
Niels van Best
Monique Mommers
John Penders
John Penders
Benno Schwikowski
Kristel Van Steen
Kristel Van Steen
author_facet Behnam Yousefi
Behnam Yousefi
Behnam Yousefi
Federico Melograna
Gianluca Galazzo
Niels van Best
Niels van Best
Monique Mommers
John Penders
John Penders
Benno Schwikowski
Kristel Van Steen
Kristel Van Steen
author_sort Behnam Yousefi
collection DOAJ
description Longitudinal analysis of multivariate individual-specific microbiome profiles over time or across conditions remains dauntin. Most statistical tools and methods that are available to study microbiomes are based on cross-sectional data. Over the past few years, several attempts have been made to model the dynamics of bacterial species over time or across conditions. However, the field needs novel views on handling microbial interactions in temporal analyses. This study proposes a novel data analysis framework, MNDA, that combines representation learning and individual-specific microbial co-occurrence networks to uncover taxon neighborhood dynamics. As a use case, we consider a cohort of newborns with microbiomes available at 6 and 9 months after birth, and extraneous data available on the mode of delivery and diet changes between the considered time points. Our results show that prediction models for these extraneous outcomes based on an MNDA measure of local neighborhood dynamics for each taxon outperform traditional prediction models solely based on individual-specific microbial abundances. Furthermore, our results show that unsupervised similarity analysis of newborns in the study, again using the notion of a taxon's dynamic neighborhood derived from time-matched individual-specific microbial networks, can reveal different subpopulations of individuals, compared to standard microbiome-based clustering, with potential relevance to clinical practice. This study highlights the complementarity of microbial interactions and abundances in downstream analyses and opens new avenues to personalized prediction or stratified medicine with temporal microbiome data.
first_indexed 2024-04-09T12:38:34Z
format Article
id doaj.art-4f33ab49af4f4d438e350ac234b65a30
institution Directory Open Access Journal
issn 1664-302X
language English
last_indexed 2024-04-09T12:38:34Z
publishDate 2023-05-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Microbiology
spelling doaj.art-4f33ab49af4f4d438e350ac234b65a302023-05-15T04:52:27ZengFrontiers Media S.A.Frontiers in Microbiology1664-302X2023-05-011410.3389/fmicb.2023.11703911170391Capturing the dynamics of microbial interactions through individual-specific networksBehnam Yousefi0Behnam Yousefi1Behnam Yousefi2Federico Melograna3Gianluca Galazzo4Niels van Best5Niels van Best6Monique Mommers7John Penders8John Penders9Benno Schwikowski10Kristel Van Steen11Kristel Van Steen12Computational Systems Biomedicine Lab, Institut Pasteur, University Paris City, Paris, FranceÉcole Doctorale Complexite du vivant, Sorbonne University, Paris, FranceBIO3—Laboratory for Systems Medicine, Department of Human Genetics, Katholieke Universiteit Leuven, Leuven, BelgiumBIO3—Laboratory for Systems Medicine, Department of Human Genetics, Katholieke Universiteit Leuven, Leuven, BelgiumDepartment of Medical Microbiology, Infectious Diseases and Infection Prevention, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, NetherlandsDepartment of Medical Microbiology, Infectious Diseases and Infection Prevention, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, NetherlandsInstitute of Medical Microbiology, Rhine-Westphalia Technical University of Aachen, RWTH University, Aachen, GermanyDepartment of Epidemiology, Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, NetherlandsDepartment of Medical Microbiology, Infectious Diseases and Infection Prevention, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, NetherlandsDepartment of Medical Microbiology, Infectious Diseases and Infection Prevention, Care and Public Health Research Institute (CAPHRI), Maastricht University Medical Center+, Maastricht, NetherlandsComputational Systems Biomedicine Lab, Institut Pasteur, University Paris City, Paris, FranceBIO3—Laboratory for Systems Medicine, Department of Human Genetics, Katholieke Universiteit Leuven, Leuven, BelgiumBIO3—Laboratory for Systems Genetics, GIGA-R Medical Genomics, University of Lièvzge, Liège, BelgiumLongitudinal analysis of multivariate individual-specific microbiome profiles over time or across conditions remains dauntin. Most statistical tools and methods that are available to study microbiomes are based on cross-sectional data. Over the past few years, several attempts have been made to model the dynamics of bacterial species over time or across conditions. However, the field needs novel views on handling microbial interactions in temporal analyses. This study proposes a novel data analysis framework, MNDA, that combines representation learning and individual-specific microbial co-occurrence networks to uncover taxon neighborhood dynamics. As a use case, we consider a cohort of newborns with microbiomes available at 6 and 9 months after birth, and extraneous data available on the mode of delivery and diet changes between the considered time points. Our results show that prediction models for these extraneous outcomes based on an MNDA measure of local neighborhood dynamics for each taxon outperform traditional prediction models solely based on individual-specific microbial abundances. Furthermore, our results show that unsupervised similarity analysis of newborns in the study, again using the notion of a taxon's dynamic neighborhood derived from time-matched individual-specific microbial networks, can reveal different subpopulations of individuals, compared to standard microbiome-based clustering, with potential relevance to clinical practice. This study highlights the complementarity of microbial interactions and abundances in downstream analyses and opens new avenues to personalized prediction or stratified medicine with temporal microbiome data.https://www.frontiersin.org/articles/10.3389/fmicb.2023.1170391/fullmicrobial neighborhood dynamicslongitudinal microbiome analysisnetwork representation learningindividual-specific networksencoder-decoder neural network
spellingShingle Behnam Yousefi
Behnam Yousefi
Behnam Yousefi
Federico Melograna
Gianluca Galazzo
Niels van Best
Niels van Best
Monique Mommers
John Penders
John Penders
Benno Schwikowski
Kristel Van Steen
Kristel Van Steen
Capturing the dynamics of microbial interactions through individual-specific networks
Frontiers in Microbiology
microbial neighborhood dynamics
longitudinal microbiome analysis
network representation learning
individual-specific networks
encoder-decoder neural network
title Capturing the dynamics of microbial interactions through individual-specific networks
title_full Capturing the dynamics of microbial interactions through individual-specific networks
title_fullStr Capturing the dynamics of microbial interactions through individual-specific networks
title_full_unstemmed Capturing the dynamics of microbial interactions through individual-specific networks
title_short Capturing the dynamics of microbial interactions through individual-specific networks
title_sort capturing the dynamics of microbial interactions through individual specific networks
topic microbial neighborhood dynamics
longitudinal microbiome analysis
network representation learning
individual-specific networks
encoder-decoder neural network
url https://www.frontiersin.org/articles/10.3389/fmicb.2023.1170391/full
work_keys_str_mv AT behnamyousefi capturingthedynamicsofmicrobialinteractionsthroughindividualspecificnetworks
AT behnamyousefi capturingthedynamicsofmicrobialinteractionsthroughindividualspecificnetworks
AT behnamyousefi capturingthedynamicsofmicrobialinteractionsthroughindividualspecificnetworks
AT federicomelograna capturingthedynamicsofmicrobialinteractionsthroughindividualspecificnetworks
AT gianlucagalazzo capturingthedynamicsofmicrobialinteractionsthroughindividualspecificnetworks
AT nielsvanbest capturingthedynamicsofmicrobialinteractionsthroughindividualspecificnetworks
AT nielsvanbest capturingthedynamicsofmicrobialinteractionsthroughindividualspecificnetworks
AT moniquemommers capturingthedynamicsofmicrobialinteractionsthroughindividualspecificnetworks
AT johnpenders capturingthedynamicsofmicrobialinteractionsthroughindividualspecificnetworks
AT johnpenders capturingthedynamicsofmicrobialinteractionsthroughindividualspecificnetworks
AT bennoschwikowski capturingthedynamicsofmicrobialinteractionsthroughindividualspecificnetworks
AT kristelvansteen capturingthedynamicsofmicrobialinteractionsthroughindividualspecificnetworks
AT kristelvansteen capturingthedynamicsofmicrobialinteractionsthroughindividualspecificnetworks